Targeting the Gastrointestinal Tract to Develop Novel Therapies for HIV RK Reeves1, A Burgener2,3,4 and NR Klatt5,6 Despite the use of antiretroviral therapy (ART), which delays and/or prevents AIDS pathogenesis, human immunodeficiency virus (HIV)infected individuals continue to face increased morbidities and mortality rates compared with uninfected individuals. Gastrointestinal (GI) mucosal dysfunction is a key feature of HIV infection, and is associated with mortality. In this study, we review current knowledge about mucosal dysfunction in HIV infection, and describe potential avenues for therapeutic targets to enhance mucosal function and decrease morbidities and mortalities in HIV-infected individuals. INTRODUCTION HIV infection in the ART era

With the recent optimization of antiretroviral drugs, most motivated human immunodeficiency virus (HIV)-infected patients with access to antiretroviral therapy (ART) can achieve durable and perhaps life-long viral suppression. Although these drugs improve quality of life, prevent AIDS, and reduce overall mortality, they do not fully restore health. A cure is not yet available, and, despite suppression of viremia with ART, individuals still have increased morbidity and mortality rates compared with uninfected persons.1–3 In addition, immune reconstitution with treatment is often incomplete even after many years of viral suppression.4 As compared to age-matched uninfected adults, ARTtreated HIV-infected adults have a higher risk of developing non-AIDS-related nonimmunological diseases, including cardiovascular disease, cancer, kidney disease, liver disease, neurologic disease, and bone disease.4,5 There are several factors that affect increased disease, such as CD4 count and viral load at onset of ART, immune reconstitution after ART, and mucosal dysfunction.4,5 Importantly, chronic mucosal dysfunction and lack of immune restoration in the gastrointestinal (GI) tract predicts and contributes to this excess risk of morbidity and mortality,1–3,6,7 characterized by epithelial barrier breaches, translocation of luminal microbes and microbial products, dysbiosis of the gut microbiome, and cellular immune dysfunction. In this review, we will describe mechanisms underlying mucosal dysfunction and potential avenues for novel therapeutic interventions, also summarized in Figure 1.

Barrier dysfunction and microbial factors

During HIV infection, focal breaches in the tight epithelial barrier of the GI tract occur, which allows microbes and microbialderived products to translocate across the barrier.8–10 This microbial translocation can directly induce inflammation by both direct stimulation of immune cells as well as by altering inflammatory pathways, such as tryptophan catabolism, and likely other consequences, such as hyperactivation and recruitment of innate immune cells. Furthermore, both microbial translocation1–3 and barrier damage6,7 have been found to be associated with morbidities and mortality in HIV infection. Although the exact mechanisms underlying barrier dysfunction are unclear, recent studies suggest that dysbiosis of the microbiome during HIV infection may play a role. Indeed, HIV infection induces alterations in the microbiome that leads to increased adherent and inflammatory microbes, such as Proteobacteria and Prevotella spp., along with loss of beneficial bacteria, such as Bacteroides spp.11–14 The result of this dysbiosis is increased inflammation, via mechanisms such as increased bacterial adherence to the epithelial barrier, increased tryptophan catabolism, and induction of inflammatory immune responses.11–14 However, the mechanisms underlying microbial dysbiosis in HIV infection have not been elucidated. The ability to improve mucosal dysfunction by targeting or preventing microbial translocation, dysbiosis of the microbiome, and/or barrier damage could potentially decrease morbidities and mortality in HIV infection. Microbial translocation has been targeted via therapeutics, such as sevelamer, which binds microbial lipopolysaccharide.15 Although this treatment in simian immunodeficiency virus (SIV)-infected macaques (a simian model of

1

Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; 2National Laboratory for HIV Immunology, Public Health Agency of Canada, Winnipeg, Canada; 3Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada; 4Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Sweden; 5Department of Pharmaceutics, University of Washington, Seattle, Washington, USA; 6Washington National Primate Research Center, Seattle, Washington, USA. Correspondence: NR Klatt ([email protected]) Received 19 June 2015; accepted 10 July 2015; advance online publication 14 July 2015. doi:10.1002/cpt.186

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 98 NUMBER 4 | OCTOBER 2015

381

Figure 1 Potential targets for intervention to improve mucosal function in HIV infection.

HIV/AIDS) resulted in decreased microbial translocation and biomarkers of mortality, it was only demonstrated to be effective if given daily during acute infection.15 Given the difficulty in identifying individuals in acute infection, it is unlikely that sevelamer would be a reasonable therapeutic option if this is not shown to be effective in chronic, treated infection. Sevelamer was also given to HIV-infected individuals in a clinical trial, and although it did not show effects in depressing microbial translocation, potential cardiovascular benefits were observed.16 In another study, HIV-infected individuals with poor immune reconstitution were given hyperimmune bovine colostrum containing anti-lipopolysaccharide immunoglobulin.17 Although positive effects were observed on CD41 cell counts, there was no decrease in biomarkers of microbial translocation.17 Further studies with novel treatment strategies that may block lipopolysaccharide activity or enhance clearance of microbial products in circulation will be crucial. Targeting the microbiome directly may be an alternative approach to decrease mucosal damage and microbial translocation. One potential avenue is by enhancing the microbiome with probiotic (beneficial bacteria) treatment. A recent study found that supplementing ART with probiotics in chronically SIVinfected macaques resulted in enhanced GI tract CD41 T cell frequency and function, and decreased fibrosis, which could be highly beneficial to health in HIV-infected individuals.18 In addition, probiotics demonstrated decreased microbial translocation, which persisted after discontinuation in HIV-infected individuals.19 As such, probiotic therapy is beginning in clinical trials in 382

HIV-infected subjects through the AIDS clinical trials group. A more dramatic possibility of altering the microbiome is through fecal material transplantation (FMT). FMT is a procedure whereby full fecal material (typically from a screened donor) is transplanted in a patient through either an endoscope via the upper intestine or colonoscopy via the lower intestine. FMT is highly effective in improving the microbiome and curing disease associated with pathogenic bacteria, such as Clostridium difficile and vancomycin-resistant Enterococcus, as well inflammatory bowel disease, all of which cause chronic and severe diarrheal disease and have been effectively cured by FMT treatment.20,21 Given the extremely positive outcomes demonstrated in other GI ailments after FMT with limited side effects, this could be an ideal treatment option aimed at enhancing mucosal function in HIV-infected individuals. Finally, the epithelial barrier is likely damaged by inflammatory responses via activated immune cells in the GI tract. Below we will discuss cellular immunity and potential mechanisms and intervention strategies to enhance barrier function via altering mucosal immunity. Homeostatic cells in mucosal tissues

Multiple lymphocyte subpopulations contribute to regulation of the gut microenvironment and disruption of this delicate balance can result in gut barrier breakdown and subsequent microbial translocation. A prime axis of regulation is mediated by the overlapping functions of T helper cells that produce the homeostatic cytokines interleukin 17 (IL-17) or IL-22 (TH17 or TH22, VOLUME 98 NUMBER 4 | OCTOBER 2015 | www.wileyonlinelibrary/cpt

respectively) and the recently described, innate lymphoid cell type 3 (ILC3), all of which are particularly relevant to HIV because of their general restriction to mucosal sites.22–29 Through production of IL-17 and IL-22, these cells play critical roles in maintaining mucosal epithelial integrity, tissue remodeling and repair, and defense against intestinal pathogens. ILC3 have even been directly shown to promote anatomic containment of mucosal bacteria.30 During HIV disease and experimental SIV infection of macaques, all three cell types are depleted numerically, both by direct infection of T cells and inflammation-induced apoptosis, and potentially other unknown mechanisms, such as phenotype skewing. Depletion occurs early after infection, is maintained throughout disease, and only moderate cell recovery is observed after ART.31–33 In addition to cellular depletion, IL17 and IL-22 production are specifically suppressed and cells are skewed toward development of T regulatory cells or an inflammatory cytotoxic phenotype that is likely to further exacerbate disease.27,32–34 Indeed, loss of IL-17/22 production in SIV-infected macaques is directly associated with loss of epithelial integrity in the gut.34 Tryptophan catabolites, IL-2, IL-12, and IL-15, all of which are upregulated in the HIV/SIV-infected gut, antagonize RORgt and RORA (the transcription factors for TH17, TH22, and ILC3), promote cell apoptosis, and have all been directly implicated in depletion of these cells.27,32,33 The exquisite sensitivity of TH17, TH22, and ILC3 to changes in inflammatory mediators in the gut milieu suggests that targeting these HIV/ SIV-induced factors could be a meaningful approach to reverse cell depletion. Adiponectin and aldosterone therapy have both been used to increase total IL-17 and the frequency of TH17 cells.35,36 Ustekinumab (Stelara) and apilimod are potent IL-12 inhibitors used in patients with psoriasis,37 but also can inhibit IL-23, an important growth factor for TH17 and ILC3, suggesting combinatorial therapies could be a beneficial approach. JAK1 inhibitors have been shown to more specifically block IL-12 secretion from dendritic cells and could be a more directed target.38 Similarly, anti-IL-15 therapies have shown promise in treating autoimmune conditions and celiac disease,39,40 and small molecule inhibitors aimed at blocking the tryptophan catabolism pathway have also proven beneficial at decreasing inflammation and clinical outcomes in cancer therapy.41 Any combination of these therapies aimed at blocking HIV/SIV-induced inflammatory mediators could aid in restoring TH17, TH22, and ILC3 numbers and thus increase total gut IL-17 and IL-22 production and have a net stabilizing effect on the mucosal barrier. Inflammatory cells in mucosal tissues

Although local TH17, TH22, and ILC3 generally have a positive impact on maintaining gut homeostasis, HIV/SIV-induced inflammation can also attract hyperactivated immune cell populations that are normally less frequent or absent from the gut. Some notable innate effector cells that can be recruited to the gut during infection are natural killer (NK) cells, inflammatory dendritic cells, including plasmacytoid dendritic cells (pDCs) and myeloid dendritic cells (mDCs), and neutrophils. Under normal conditions, NK cells are relatively infrequent in the gut mucosa but are important for defense against various mucosal pathogens. However, in both CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 98 NUMBER 4 | OCTOBER 2015

HIV and SIV infection, the normal homing of NK cells in and out of lymph nodes is interrupted with an overall shift toward guttrafficking where they may accumulate in a hypercytotoxic state.27,42–46 Although increased NK cells could aid clearance of infected cells, overactivation can also result in nonspecific and bystander killing of immune cells.47–49 To this end, multiple strategies have been used to deplete NK cells in SIV-infected rhesus macaques, the most effective of which being JAK3 inhibitors.50–52 However, despite potential benefits to the gut, NK cell depletion in SIV-infected macaques has generally resulted in increased virus replication. Alternatively, NK cells observed in chronic HIV and SIV infection are significantly dysfunctional and may be unable to perform both regulatory functions and defense against opportunistic pathogens. Therefore, strategies not to deplete NK cells, but rather restore normal function may be a more appropriate strategy depending on clinical presentation. Altered dendritic cells in the gut could vastly affect overall immunity and regulation of the mucosal barrier given the important role of these cells in priming and inducing immunity. The pDCs are the primary IFN-a-producing cells in the body and, although they serve as a first line of defense against viral infection,53 accumulation of pDCs in the mucosa during HIV/SIV infection is associated with elevated levels of IFN-a in the gut, as well as increased immune activation, apoptosis, target cell recruitment, and dysregulation of anti-HIV/SIV T cell responses.54–58 Initial studies in acute SIV infection have shown that blocking IFN-a production by pDCs using TLR7/9 antagonists does not diminish immune activation, but this approach provided incomplete inhibition and was not specific to pDCs.59 Although specifically depleting pDCs in vivo is an attractive target, no effective approach has been identified outside of murine models. Alternatively, direct blocking of IFN-a with monoclonal antibody therapies, such as sifalimumab, have been effective in treatment of lupus, another disease with IFN-a-induced pathogenesis.60 AntiIFN-a vaccination has also been tried in patients with HIV, and, while generally well tolerated, the clinical benefits were very modest.61 Chloroquine has also been used to reduce IFN-a through its ability to inhibit endosomal acidification, and indeed these approaches significantly reduced immune activation and measures of microbial translocation.62 Collectively, these data suggest that, although pDCs and IFN-a might be beneficial if properly harnessed during transmission or acute disease,59 their overaccumulation in the gut during chronic infection is generally deleterious. As such, combination approaches to regulate pDC numbers and/or production of IFN-a in the gut could be attractive as a modality to improve disease outcomes. The mDCs are typically thought to be more important in priming and inducing antigen-specific immunity, particularly by presenting antigen to T cells. In HIV infection, dysbiosis of the microbiome has been demonstrated to result in altered mDCs, which in turn is associated with increased T cell activation both in mucosal and systemic sites.12,13 Although targeting mDCs could be dangerous in that reduction of function of these cells could result in decreased antigen-specific immunity, targeting the microbiome (as described above) may decrease the hyperactivation and alteration of mDCs in the GI tract and thus decrease inflammation. 383

Another potential consequence of dysbiosis of the microbiome is neutrophil accumulation in the GI tract, which has been demonstrated in HIV infected individuals.6 Given the propensity for neutrophils to produce high levels of inflammatory factors, such as proteases, which can directly cause breakdown of epithelial integrity, accumulation and hyperactivation of these cells because of microbial translocation and dysbiosis may be a direct source of mucosal damage. Neutrophils proteases, which include serine-type (cathepsin, elastase, proteinase 3) and matrix metalloproteinases,63 although secreted to combat invading microorganisms, can paradoxically damage host tissues by degrading structural proteins of mucosal surfaces.63,64 Serine proteases and matrix metalloproteinases cleave and redistribute cell–cell junction and adhesion proteins, including those of the EMC, and change endothelial and epithelial barrier integrity.65 Occludens, fibrins, collagen, cadherins, and others are all targets for matrix metalloproteinases, whereas neutrophil elastase targets EMC proteins, such as elastin and collagen.66 Antiproteases, such as serpins (serine protease inhibitors) and tissue inhibitor of metalloproteinases regulate their activity, but skewing of the protease-antiprotease balance is associated with barrier disruption, such as in asthma and chronic obstructive pulmonary disease,67 and the inability to clear bacterial infections and mucosal tissue damage.68 Sustained release of matrix metalloproteinases by accumulated neutrophils impedes repair of the intestinal epithelium by generation of cleavage products from surface of neutrophil-associated junctional adhesion proteins.69 In addition, a relationship between neutrophil proteases and mucosal barrier disruption in the female reproductive tract has been observed and proposed as a mechanism of increased HIV acquisition risk.70 Indeed, elevated serine antiproteases (serpins, elafin) are strongly linked with protection from HIV acquisition.71–74 Therefore, this demonstrates a link between the neutrophil proteases and mucosal barrier disruption in the context of HIV, and by extension these may serve as therapeutic potential for intervention in gut function in HIV infected individuals. CONCLUSION

Taken together, there are several potential avenues for therapeutic intervention in the GI tract during HIV infection that may benefit health (Figure 1). Currently, many studies are under way to address inflammation in HIV-infected individuals in the ART era, and, given the increased understanding of many mechanisms underlying GI dysfunction, microbial translocation and inflammation in the context of HIV infection has provided several novel pathways for intervention strategies. Ideally, supplementing ART with an anti-inflammatory or GI-enhancing regimen will be without side effects and simple to ensure patient adherence and outweigh the risks compared to the benefits. Overall, further understanding of the mechanisms underlying GI dysfunction and identifying specific targets for novel interventions should be highly prioritized to decrease morbidities and mortality in HIV infection. ACKNOWLEDGMENT The authors thank Laura Romas for her assistance in the creation of Figure 1. 384

CONFLICT OF INTEREST The authors declared no conflict of interest.

C 2015 American Society for Clinical Pharmacology and Therapeutics V

1.

2.

3. 4.

5.

6. 7.

8.

9.

10.

11.

12.

13.

14. 15.

16.

17.

18.

19.

20.

Sandler, N.G. et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J. Infect. Dis. 203, 780–790 (2011). Rodger, A.J. et al. Activation and coagulation biomarkers are independent predictors of the development of opportunistic disease in patients with HIV infection. J. Infect. Dis. 200, 973– 983 (2009). Kuller, L.H. et al. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 5, e203 (2008). Klatt, N.R., Chomont, N., Douek, D.C. & Deeks, S.G. Immune activation and HIV persistence: implications for curative approaches to HIV infection. Immunol. Rev. 254, 326–342 (2013). Lederman, M.M., Funderburg, N.T., Sekaly, R.P., Klatt, N.R. & Hunt, P.W. Residual immune dysregulation syndrome in treated HIV infection. Adv. Immunol. 119, 51–83 (2013). Somsouk, M. et al. Gut epithelial barrier and systemic inflammation during chronic HIV infection. AIDS 29, 43–51 (2015). Hunt, P.W. et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J. Infect. Dis. 210, 1228–1238 (2014). Klatt, N.R., Funderburg, N.T. & Brenchley, J.M. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 21, 6–13 (2013). Brenchley, J.M. et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 12, 1365– 1371 (2006). Estes, J.D. et al. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLoS Pathog. 6, e1001052 (2010). Vujkovic-Cvijin, I. et al. Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci. Transl. Med. 5, 193ra91 (2013). Dillon, S.M. et al. Gut dendritic cell activation links an altered colonic microbiome to mucosal and systemic T-cell activation in untreated HIV-1 infection. Mucosal Immunol. (2015); e-pub ahead of print. Dillon, S.M. et al. An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol. 7, 983–994 (2014). Klase, Z. et al. Dysbiotic bacteria translocate in progressive SIV infection. Mucosal Immunol. (2015); e-pub ahead of print. Kristoff, J. et al. Early microbial translocation blockade reduces SIVmediated inflammation and viral replication. J. Clin. Invest. 124, 2802–2806 (2014). Sandler, N.G. et al. Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, lowdensity lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection. J. Infect. Dis. 210, 1549–1554 (2014). Byakwaga, H. et al. Intensification of antiretroviral therapy with raltegravir or addition of hyperimmune bovine colostrum in HIVinfected patients with suboptimal CD41 T-cell response: a randomized controlled trial. J. Infect. Dis. 204, 1532–1540 (2011). Klatt, N.R. et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques. J. Clin. Invest. 123, 903–907 (2013). Villar-Garcıa, J. et al. Effect of probiotics (Saccharomyces boulardii) on microbial translocation and inflammation in HIV-treated patients: a double-blind, randomized, placebo-controlled trial. J. Acquir. Immune Defic. Syndr. 68, 256–263 (2015). Mattila, E. et al. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology 142, 490–496 (2012). VOLUME 98 NUMBER 4 | OCTOBER 2015 | www.wileyonlinelibrary/cpt

21. Smits, L.P., Bouter, K.E., de Vos, W.M., Borody, T.J. & Nieuwdorp, M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology 145, 946–953 (2013). 22. Spits, H. & Di Santo, J.P. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nat. Immunol. 12, 21–27 (2011). 23. Satoh-Takayama, N. et al. Microbial flora drives interleukin 22 production in intestinal NKp461 cells that provide innate mucosal immune defense. Immunity 29, 958–970 (2008). 24. Zheng, Y. et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat. Med. 14, 282–289 (2008). 25. Cella, M. et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 457, 722– 725 (2009). 26. Takatori, H. et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J. Exp. Med. 206, 35–41 (2009). 27. Reeves, R.K. et al. Gut inflammation and indoleamine deoxygenase inhibit IL-17 production and promote cytotoxic potential in NKp441 mucosal NK cells during SIV infection. Blood 118, 3321–3330 (2011). 28. Raffatellu, M. et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat. Med. 14, 421–428 (2008). 29. Mucida, D. & Salek-Ardakani, S. Regulation of TH17 cells in the mucosal surfaces. J. Allergy Clin. Immunol. 123, 997–1003 (2009). 30. Sonnenberg, G.F., Monticelli, L.A., Elloso, M.M., Fouser, L.A. & Artis, D. CD4(1) lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011). 31. Schuetz, A. et al. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation. PLoS Pathog. 10, e1004543 (2014). 32. Li, H. et al. Hypercytotoxicity and rapid loss of NKp441 innate lymphoid cells during acute SIV infection. PLoS Pathog. 10, e1004551 (2014). 33. Favre, D. et al. Tryptophan catabolism by indoleamine 2,3dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Sci. Transl. Med. 2, 32ra36 (2010). 34. Favre, D. et al. Critical loss of the balance between Th17 and T regulatory cell populations in pathogenic SIV infection. PLoS Pathog. 5, e1000295 (2009). 35. Jung, M.Y., Kim, H.S., Hong, H.J., Youn, B.S. & Kim, T.S. Adiponectin induces dendritic cell activation via PLCg/JNK/NF-jB pathways, leading to Th1 and Th17 polarization. J. Immunol. 188, 2592–2601 (2012). 36. Herrada, A.A. et al. Aldosterone promotes autoimmune damage by enhancing Th17-mediated immunity. J. Immunol. 184, 191–202 (2010). 37. Brezinski, E.A. & Armstrong, A.W. An evidence-based review of the mechanism of action, efficacy, and safety of biologic therapies in the treatment of psoriasis and psoriatic arthritis. Curr. Med. Chem. 22, 1930–1942 (2015). 38. Wagner, A.H. et al. JAK1/STAT3 activation directly inhibits IL-12 production in dendritic cells by preventing CDK9/P-TEFb recruitment to the p35 promoter. Biochem. Pharmacol. 96, 52–64 (2015). 39. Baslund, B. et al. Targeting interleukin-15 in patients with rheumatoid arthritis: a proof-of-concept study. Arthritis Rheum. 52, 2686–2692 (2005). 40. Benahmed, M. et al. Inhibition of TGF-beta signaling by IL-15: a new role for IL-15 in the loss of immune homeostasis in celiac disease. Gastroenterology 132, 994–1008 (2007). 41. Prendergast, G.C. et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol. Immunother. 63, 721–735 (2014). 42. Reeves, R.K., Evans, T.I., Gillis, J. & Johnson, R.P. Simian immunodeficiency virus infection induces expansion of alpha4beta71 and cytotoxic CD561 NK cells. J. Virol. 84, 8959– 8963 (2010). 43. Sips, M. et al. Altered distribution of mucosal NK cells during HIV infection. Mucosal Immunol. 5, 30–40 (2012). 44. Mela, C.M., Steel, A., Lindsay, J., Gazzard, B.G., Gotch, F.M. & Goodier, M.R. Depletion of natural killer cells in the colonic lamina propria of viraemic HIV-1-infected individuals. AIDS 21, 2177–2182 (2007). CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 98 NUMBER 4 | OCTOBER 2015

45. Naranbhai, V., Altfeld, M., Karim, S.S., Ndung’u, T., Karim, Q.A. & Carr, W.H. Changes in natural killer cell activation and function during primary HIV-1 infection. PLoS One 8, e53251 (2013). 46. Hong, H.S. et al. Loss of CCR7 expression on CD56(bright) NK cells is associated with a CD56(dim)CD161 NK cell-like phenotype and correlates with HIV viral load. PLoS One 7, e44820 (2012). 47. Kottilil, S. et al. Innate immunity in HIV infection: enhanced susceptibility to CD95-mediated natural killer cell death and turnover induced by HIV viremia. J. Acquir. Immune Defic. Syndr. 46, 151–159 (2007). 48. Kramski, M., Parsons, M.S., Stratov, I. & Kent, S.J. HIV-specific antibody immunity mediated through NK cells and monocytes. Curr. HIV Res. 11, 388–406 (2013). 49. Richard, J., Pham, T.N., Ishizaka, Y. & Cohen, E.A. Viral protein R upregulates expression of ULBP2 on uninfected bystander cells during HIV-1 infection of primary CD41 T lymphocytes. Virology. 443, 248–256 (2013). 50. Choi, E.I., Reimann, K.A. & Letvin, N.L. In vivo natural killer cell depletion during primary simian immunodeficiency virus infection in rhesus monkeys. J. Virol. 82, 6758–6761 (2008). 51. Takahashi, Y. et al. In vivo administration of a JAK3 inhibitor during acute SIV infection leads to significant increases in viral load during chronic infection. PLoS Pathog. 10, e1003929 (2014). 52. Okoye, A. et al. Anti-IL15 administration induces increases in CD4 central memory T cells in SIV infected rhesus macaques. 28th Annual Symposium on Nonhuman Primate Models for AIDS; October 19–22, 2010; New Orleans, Louisianna. 53. Siegal, F.P. et al. The nature of the principal type 1 interferonproducing cells in human blood. Science 284, 1835–1837 (1999). 54. Lehmann, C. et al. Longitudinal analysis of distribution and function of plasmacytoid dendritic cells in peripheral blood and gut mucosa of HIV infected patients. J. Infect. Dis. 209, 940–949 (2014). 55. Reeves, R.K. et al. SIV infection induces accumulation of plasmacytoid dendritic cells in the gut mucosa. J. Infect. Dis. 206, 1462–1468 (2012). 56. Li, H., Gillis, J., Johnson, R.P. & Reeves, R.K. Multi-functional plasmacytoid dendritic cells redistribute to gut tissues during simian immunodeficiency virus infection. Immunology 140, 244–249 (2013). 57. Li, H., Goepfert, P. & Reeves, R.K. Short communication: plasmacytoid dendritic cells from HIV-1 elite controllers maintain a gut-homing phenotype associated with immune activation. AIDS Res. Hum. Retroviruses 30, 1213–1215 (2014). 58. Boasso, A. et al. Overactivation of plasmacytoid dendritic cells inhibits antiviral T-cell responses: a model for HIV immunopathogenesis. Blood 118, 5152–5162 (2011). 59. Sandler, N.G. et al. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature 511, 601–605 (2014). 60. Kirou, K.A. & Gkrouzman, E. Anti-interferon alpha treatment in SLE. Clin. Immunol. 148, 303–312 (2013). 61. Gringeri, A. et al. Active anti-interferon-alpha immunization: a European-Israeli, randomized, double-blind, placebo-controlled clinical trial in 242 HIV-1--infected patients (the EURIS study). J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 20, 358–370 (1999). 62. Murray, S.M. et al. Reduction of immune activation with chloroquine therapy during chronic HIV infection. J. Virol. 84, 12082–12086 (2010). 63. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 13, 159–175 (2013). €no €nen, E., Luukkonen, N. & Uitto, V.J. Human 64. Gursoy, U.K., Ko neutrophil defensins and their effect on epithelial cells. J. Periodontol. 84, 126–133 (2013). 65. Vermeer, P.D. et al. MMP9 modulates tight junction integrity and cell viability in human airway epithelia. Am. J. Physiol. Lung Cell Mol. Physiol. 296, L751–L762 (2009). pez-Boado, Y.S. Matrix 66. Parks, W.C., Wilson, C.L. & Lo metalloproteinases as modulators of inflammation and innate immunity. Nat. Rev. Immunol. 4, 617–629 (2004). 67. Barnes, P.J. New treatments for COPD. Nat. Rev. Drug Discov. 1, 437–446 (2002). 68. Benarafa, C., Priebe, G.P. & Remold-O’Donnell, E. The neutrophil serine protease inhibitor serpinb1 preserves lung defense functions 385

in Pseudomonas aeruginosa infection. J. Exp. Med. 204, 1901–1909 (2007). 69. Weber, D.A. et al. Neutrophil-derived JAML inhibits repair of intestinal epithelial injury during acute inflammation. Mucosal Immunol. 7, 1221–1232 (2014). 70. Arnold, K.B. et al. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV susceptible target cells. Mucosal Immunol. (2015); e-pub ahead of print. 71. Lajoie, J. et al. A distinct cytokine and chemokine profile at the genital mucosa is associated with HIV-1 protection among HIV-

386

exposed seronegative commercial sex workers. Mucosal Immunol. 5, 277–287 (2012). 72. Burgener, A. et al. Comprehensive proteomic study identifies serpin and cystatin antiproteases as novel correlates of HIV-1 resistance in the cervicovaginal mucosa of female sex workers. J. Proteome Res. 10, 5139–5149 (2011). 73. Burgener, A. et al. Identification of differentially expressed proteins in the cervical mucosa of HIV-1-resistant sex workers. J. Proteome Res. 7, 4446–4454 (2008). 74. Aboud, L., Ball, T.B., Tjernlund, A. & Burgener, A. The role of serpin and cystatin antiproteases in mucosal innate immunity and their defense against HIV. Am. J. Reprod. Immunol. 71, 12–23 (2013).

VOLUME 98 NUMBER 4 | OCTOBER 2015 | www.wileyonlinelibrary/cpt

Targeting the gastrointestinal tract to develop novel therapies for HIV.

Despite the use of antiretroviral therapy (ART), which delays and/or prevents AIDS pathogenesis, human immunodeficiency virus (HIV)-infected individua...
3MB Sizes 3 Downloads 8 Views